Targeting METTL14 and RNA Methylation to Treat Osteosarcoma.
Lili Ren,Xiaobo Li,Rui Su
DOI: https://doi.org/10.1016/j.ebiom.2022.104190
IF: 11.205
2022-01-01
EBioMedicine
Abstract:Osteosarcoma (OS) is one of the most common and aggressive primary bone malignancies in both adults and children. During the past 4 decades, the substantial improvements in surgery and neoadjuvant chemotherapy have significantly increased the survival rate of OS patients. However, with currently available treatment regimens, more than 80% of the OS patients with metastasis and/or relapse do not survive over 5 years,1Gill J Gorlick R. Advancing therapy for osteosarcoma.Nat Rev Clin Oncol. 2021; 18: 609-624https://doi.org/10.1038/s41571-021-00519-8Crossref PubMed Scopus (81) Google Scholar which underscores the unmet medical need to discover novel effective therapeutic target(s). The poor understanding of cellular and molecular mechanisms during osteosarcomagenesis has seriously prevented the development of new treatment approaches.In eukaryotes, the gene expression is strictly regulated at multiple layers, including genetics, epigenetics, and epitranscriptomics, to maintain the normal bioprocess, while dysregulation of gene expression may lead to pathogenesis and tumorigenesis. The covalent chemical modifications in RNA have been revealed since 1950s and emerged as a crucial layer to manipulate gene expression at post-transcriptional level. To date, although more than 170 distinct chemical marks have been identified in RNAs, N6-methyladenosine (m6A), methylated adenosine at the N6 position, is the most abundant internal modification in messenger RNA (mRNA). Since the initial discovery in 1974, the studies focusing on m6A modification have been stuck for several decades due to the lack of quantification and sequencing approaches. Until 2011, identification of the first m6A demethylase, fat mass and obesity-associated protein (FTO), indicates that m6A is dynamic and reversable and resurged the broad interest in RNA modifications. The m6A is always enriched in the RRACH (R=A or G; H = A, C, or U) context around stop codon region, and its machinery includes “writers”, “erasers” and “readers” that deposit, remove, and recognize it, respectively.2Shi H Wei J He C. Where, when, and how: context-dependentfundtions of RNA methylation writers, readers, and erasers.Mol Cell. 2019; 16: 640-650https://doi.org/10.1016/j.molcel.2019.04.025Summary Full Text Full Text PDF Scopus (573) Google Scholar Specifically, the m6A methylation is co-transcriptionally installed in mRNA by a multicomponent complex composed of a stable core subunit, the methyltransferase-like 3 (METTL3)/METTL14 heterodimer, and additional regulators. The m6A modification can be reversed by a-ketoglutarate (a-KG)-dependent and Fe(II)-dependent demethylases, FTO and AlkB homolog 5 (ALKBH5). The characterization of m6A readers, such as YT521-B homology (YTH) domain family 1-3 (YTHDF1-3), YTH domain containing 1-2 (YTHDC1-2), and insulin-like growth factor 2 mRNA-binding proteins 1-3 (IGF2BP1-3), has provided insightful knowledge into the underlying mechanisms of m6A-mediated gene expression regulation, including nuclear export, mRNA stability, alternative splicing, and translation efficiency. Therefore, m6A represents the best characterized RNA modification thus far. Recently, accumulating evidence have suggested that both the m6A abundance and its machinery are usually dysregulated in various cancers and such dysregulation is responsible for tumor initiation, progression, drug resistance, metastasis, and refractory.3Weng H Huang H Wu H et al.METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m6A Modification.Cell Stem Cell. 2018; 191: 191-205.e9https://doi.org/10.1016/j.stem.2017.11.016Summary Full Text Full Text PDF Scopus (527) Google Scholar More exciting, the novel small compounds targeting m6A machinery have been developed and utilized to treat tumors, especially leukemia.4Su R Dong L Li Y et al.Targeting FTO suppresses cancer stem cell maintenance and immune evasion.Cancer Cell. 2020; 38 (79-96.e11): 79-96.e11https://doi.org/10.1016/j.ccell.2020.04.017Summary Full Text Full Text PDF PubMed Scopus (205) Google Scholar However, the biological functions and molecular mechanisms of m6A and its regulators remain poorly understood in OS. It's still elusive about the role of m6A machinery during osteosarcomagenesis.In a recent issue of eBioMedicine, Li and colleagues determined the global m6A abundance in OS tumors, systemically revealed the crucial tumor-promoting role of METTL14 during OS progression and metastasis, and highlighted that METTL14 might act as a promising therapeutic target for OS treatment.5Li Hong-Bo Huang Gang Tu Jian et al.METTL14-mediated epitranscriptome modification of MN1 mRNA promote tumorigenicity and all-trans-retinoic acid resistance in osteosarcoma.EBiomedicine. 2022; 82104142Summary Full Text Full Text PDF Scopus (2) Google Scholar Here, the authors observed that the overall m6A abundance was significantly up-regulated in OS tumors in contrast to their corresponding controls, and such increased m6A modification in mRNA was mainly attributed to the robust upregulation of METTL14, but not other m6A regulators. Furthermore, the higher expression of METTL14 predicted the more adverse survival of OS patients. Via both in vitro and in vivo studies, the authors demonstrated that METTL14 played a critical oncogenic role in OS via facilitating cell growth/proliferation, promoting lung metastasis, and maintaining the stemness of cancer stem cells (CSCs). To further clarify the underlying cellular/molecular mechanism, they conducted transcriptome-wide m6A-modified RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) in OS cells upon METTL14 depletion, demonstrated Meningioma 1 (MN1) as a functionally essential and m6A-dependent downstream target of METTL14. Mechanistically, METTL14 could directly install m6A modification in MN1 mRNA, and IGF2BP2 specifically recognized the m6A marks and potentiated the mRNA stability and translation efficiency of MN1 in OS. Additionally, previous studies have supported that forced expression of MN1 conferred resistance to all-trans-retinoic acid (ATRA)-mediated anti-tumor activity in acute promyelocytic leukemia (APL),6Lo-Coco F Avvisati G Vignetti M et al.Retinoic acid and arsenic trioxide for acute promyelocytic leukemia.N Engl J Med. 2013; 369: 111-121https://doi.org/10.1056/NEJMoa1300874Crossref PubMed Scopus (970) Google Scholar while it's still underdetermined whether such effect is involved in OS. In this study, the authors also demonstrated that METTL14/m6A/IGF2BP2 axis-induced upregulation of MN could lead to the chemotherapy resistance to ATRA in OS, and suggested the potent therapeutic efficiency of ATRA in treating OS patients with endogenous low expression of MN1.Overall, this proof-of-concept article has comprehensively investigated the oncogenic role of m6A writer METTL14, elucidated its molecular mechanism via targeting MN1, and indicated that METTL14 might function as a prognostic predictor and a promising therapeutic target for OS patients. For further bench-to-bedside research, there are still a series of fundamental and clinical questions to be answered: (1) Given that several previous studies have reported the opposite (tumor-suppressor) role of METTL14 in OS,7Li Z Liu N Huang Z Wang W. METTL14 overexpression promotes osteosarcoma cell apoptosis and slows tumor progression via caspase 3 activation.Caner Manag Res. 2020; 12: 12759-12767https://doi.org/10.2147/CMAR.S284273Crossref Scopus (14) Google Scholar,8Li J Rao B Yang J et al.Dysregulated m6A-related regulators are associated with tumor metastasis and poor prognosis in osteosarcoma.Front Oncol. 2020; 10769https://doi.org/10.3389/fonc.2020.00769Crossref Scopus (39) Google Scholar further studies will be warranted to elucidate the divergent functions of m6A modification and its writer in distinct OS subtypes; (2) It's also necessary to evaluate whether METTL14 is required for the survival and growth of normal human osteoblasts; (3) ATRA is not the standard first-line chemotherapy agent to treat OS and the IC50 values of ATRA in killing OS cells are pretty high (> 50 μM), indicating ATRA might be not the ideal drug for OS therapy. It will be of significance to assess whether genetic depletion and/or pharmacological inhibition of METTL14 can sensitize OS cells to standard chemotherapy, including doxorubicin, cisplatin, and methotrexate.ContributorsLiterature search: R.L. and X.L.; Data collection R.L., X.L., and R.S.; Data interpretation: X.L. and R.S.; Writing: R.L., X.L., and R.S. All authors read and approved the final manuscript. Osteosarcoma (OS) is one of the most common and aggressive primary bone malignancies in both adults and children. During the past 4 decades, the substantial improvements in surgery and neoadjuvant chemotherapy have significantly increased the survival rate of OS patients. However, with currently available treatment regimens, more than 80% of the OS patients with metastasis and/or relapse do not survive over 5 years,1Gill J Gorlick R. Advancing therapy for osteosarcoma.Nat Rev Clin Oncol. 2021; 18: 609-624https://doi.org/10.1038/s41571-021-00519-8Crossref PubMed Scopus (81) Google Scholar which underscores the unmet medical need to discover novel effective therapeutic target(s). The poor understanding of cellular and molecular mechanisms during osteosarcomagenesis has seriously prevented the development of new treatment approaches. In eukaryotes, the gene expression is strictly regulated at multiple layers, including genetics, epigenetics, and epitranscriptomics, to maintain the normal bioprocess, while dysregulation of gene expression may lead to pathogenesis and tumorigenesis. The covalent chemical modifications in RNA have been revealed since 1950s and emerged as a crucial layer to manipulate gene expression at post-transcriptional level. To date, although more than 170 distinct chemical marks have been identified in RNAs, N6-methyladenosine (m6A), methylated adenosine at the N6 position, is the most abundant internal modification in messenger RNA (mRNA). Since the initial discovery in 1974, the studies focusing on m6A modification have been stuck for several decades due to the lack of quantification and sequencing approaches. Until 2011, identification of the first m6A demethylase, fat mass and obesity-associated protein (FTO), indicates that m6A is dynamic and reversable and resurged the broad interest in RNA modifications. The m6A is always enriched in the RRACH (R=A or G; H = A, C, or U) context around stop codon region, and its machinery includes “writers”, “erasers” and “readers” that deposit, remove, and recognize it, respectively.2Shi H Wei J He C. Where, when, and how: context-dependentfundtions of RNA methylation writers, readers, and erasers.Mol Cell. 2019; 16: 640-650https://doi.org/10.1016/j.molcel.2019.04.025Summary Full Text Full Text PDF Scopus (573) Google Scholar Specifically, the m6A methylation is co-transcriptionally installed in mRNA by a multicomponent complex composed of a stable core subunit, the methyltransferase-like 3 (METTL3)/METTL14 heterodimer, and additional regulators. The m6A modification can be reversed by a-ketoglutarate (a-KG)-dependent and Fe(II)-dependent demethylases, FTO and AlkB homolog 5 (ALKBH5). The characterization of m6A readers, such as YT521-B homology (YTH) domain family 1-3 (YTHDF1-3), YTH domain containing 1-2 (YTHDC1-2), and insulin-like growth factor 2 mRNA-binding proteins 1-3 (IGF2BP1-3), has provided insightful knowledge into the underlying mechanisms of m6A-mediated gene expression regulation, including nuclear export, mRNA stability, alternative splicing, and translation efficiency. Therefore, m6A represents the best characterized RNA modification thus far. Recently, accumulating evidence have suggested that both the m6A abundance and its machinery are usually dysregulated in various cancers and such dysregulation is responsible for tumor initiation, progression, drug resistance, metastasis, and refractory.3Weng H Huang H Wu H et al.METTL14 inhibits hematopoietic stem/progenitor differentiation and promotes leukemogenesis via mRNA m6A Modification.Cell Stem Cell. 2018; 191: 191-205.e9https://doi.org/10.1016/j.stem.2017.11.016Summary Full Text Full Text PDF Scopus (527) Google Scholar More exciting, the novel small compounds targeting m6A machinery have been developed and utilized to treat tumors, especially leukemia.4Su R Dong L Li Y et al.Targeting FTO suppresses cancer stem cell maintenance and immune evasion.Cancer Cell. 2020; 38 (79-96.e11): 79-96.e11https://doi.org/10.1016/j.ccell.2020.04.017Summary Full Text Full Text PDF PubMed Scopus (205) Google Scholar However, the biological functions and molecular mechanisms of m6A and its regulators remain poorly understood in OS. It's still elusive about the role of m6A machinery during osteosarcomagenesis. In a recent issue of eBioMedicine, Li and colleagues determined the global m6A abundance in OS tumors, systemically revealed the crucial tumor-promoting role of METTL14 during OS progression and metastasis, and highlighted that METTL14 might act as a promising therapeutic target for OS treatment.5Li Hong-Bo Huang Gang Tu Jian et al.METTL14-mediated epitranscriptome modification of MN1 mRNA promote tumorigenicity and all-trans-retinoic acid resistance in osteosarcoma.EBiomedicine. 2022; 82104142Summary Full Text Full Text PDF Scopus (2) Google Scholar Here, the authors observed that the overall m6A abundance was significantly up-regulated in OS tumors in contrast to their corresponding controls, and such increased m6A modification in mRNA was mainly attributed to the robust upregulation of METTL14, but not other m6A regulators. Furthermore, the higher expression of METTL14 predicted the more adverse survival of OS patients. Via both in vitro and in vivo studies, the authors demonstrated that METTL14 played a critical oncogenic role in OS via facilitating cell growth/proliferation, promoting lung metastasis, and maintaining the stemness of cancer stem cells (CSCs). To further clarify the underlying cellular/molecular mechanism, they conducted transcriptome-wide m6A-modified RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) in OS cells upon METTL14 depletion, demonstrated Meningioma 1 (MN1) as a functionally essential and m6A-dependent downstream target of METTL14. Mechanistically, METTL14 could directly install m6A modification in MN1 mRNA, and IGF2BP2 specifically recognized the m6A marks and potentiated the mRNA stability and translation efficiency of MN1 in OS. Additionally, previous studies have supported that forced expression of MN1 conferred resistance to all-trans-retinoic acid (ATRA)-mediated anti-tumor activity in acute promyelocytic leukemia (APL),6Lo-Coco F Avvisati G Vignetti M et al.Retinoic acid and arsenic trioxide for acute promyelocytic leukemia.N Engl J Med. 2013; 369: 111-121https://doi.org/10.1056/NEJMoa1300874Crossref PubMed Scopus (970) Google Scholar while it's still underdetermined whether such effect is involved in OS. In this study, the authors also demonstrated that METTL14/m6A/IGF2BP2 axis-induced upregulation of MN could lead to the chemotherapy resistance to ATRA in OS, and suggested the potent therapeutic efficiency of ATRA in treating OS patients with endogenous low expression of MN1. Overall, this proof-of-concept article has comprehensively investigated the oncogenic role of m6A writer METTL14, elucidated its molecular mechanism via targeting MN1, and indicated that METTL14 might function as a prognostic predictor and a promising therapeutic target for OS patients. For further bench-to-bedside research, there are still a series of fundamental and clinical questions to be answered: (1) Given that several previous studies have reported the opposite (tumor-suppressor) role of METTL14 in OS,7Li Z Liu N Huang Z Wang W. METTL14 overexpression promotes osteosarcoma cell apoptosis and slows tumor progression via caspase 3 activation.Caner Manag Res. 2020; 12: 12759-12767https://doi.org/10.2147/CMAR.S284273Crossref Scopus (14) Google Scholar,8Li J Rao B Yang J et al.Dysregulated m6A-related regulators are associated with tumor metastasis and poor prognosis in osteosarcoma.Front Oncol. 2020; 10769https://doi.org/10.3389/fonc.2020.00769Crossref Scopus (39) Google Scholar further studies will be warranted to elucidate the divergent functions of m6A modification and its writer in distinct OS subtypes; (2) It's also necessary to evaluate whether METTL14 is required for the survival and growth of normal human osteoblasts; (3) ATRA is not the standard first-line chemotherapy agent to treat OS and the IC50 values of ATRA in killing OS cells are pretty high (> 50 μM), indicating ATRA might be not the ideal drug for OS therapy. It will be of significance to assess whether genetic depletion and/or pharmacological inhibition of METTL14 can sensitize OS cells to standard chemotherapy, including doxorubicin, cisplatin, and methotrexate. ContributorsLiterature search: R.L. and X.L.; Data collection R.L., X.L., and R.S.; Data interpretation: X.L. and R.S.; Writing: R.L., X.L., and R.S. All authors read and approved the final manuscript. Literature search: R.L. and X.L.; Data collection R.L., X.L., and R.S.; Data interpretation: X.L. and R.S.; Writing: R.L., X.L., and R.S. All authors read and approved the final manuscript. The authors declare no conflict of interest. This study was supported by the HMU Marshal Initiative Funding (HMUMIF-21001, X.L.), Leukemia Research Foundation New Investigator Research Grant (R. S.), The Margaret E. Early Medical Research Trust (R.S.), and American Association for the Study of Liver Diseases (AASLD) Foundation PNC22-261362 (R.S.). METTL14-mediated epitranscriptome modification of MN1 mRNA promote tumorigenicity and all-trans-retinoic acid resistance in osteosarcomaOur study revealed that METTL14 contributes to OS progression and ATRA resistance as an m6A RNA methylase by regulating the stability and translation efficiency of MN1 and thus provides both an underlying biomarker panel for prognosis prediction in OS patients. Full-Text PDF Open Access
What problem does this paper attempt to address?
-
The M6A Methyltransferase METTL14 Promotes Myelodysplastic Syndromes Development Via PI3K/Akt Signaling Pathway
Lingxu Jiang,Xinping Zhou,Chao Hu,Chen Mei,Gaixiang Xu,Liya Ma,Yanling Ren,Li Ye,Lu Wang,Yingwan Luo,Jie Jin,Jie Sun,Hongyan Tong
DOI: https://doi.org/10.1182/blood-2022-165203
IF: 20.3
2022-01-01
Blood
Abstract:Background: RNA N6-methyladenosine (m6A) plays a critical role in normal and malignant hematopoiesis. The m6A methyltransferases METTL3 and METTL14 were reported as oncogenes in acute myeloid leukemia. Epigenetic dysregulation is a prominent biological feature of myelodysplastic syndromes (MDS), however, whether RNA methylation dysregulation exists in MDS, and what's the functions of m6A methyltransferases are largely unknown. Methods: The global m6A modification level of RNA in bone marrow was detected in MDS patients by colorimetric method, and the expression level of METTL14 in bone marrow of MDS patients was detected by q-PCR. Both gain- and loss-of-function experiments were performed to study the role of METTL14 in MDS. M6A sequencing and RNA sequencing were examined to explore the potential signaling pathways and target genes that mediated METTL14 function in MDS. Results: We detected obvious RNA m6A dysregulation in bone marrow of MDS patients compared with normal controls, and found that the up-regulation of the global m6A modification is closely related to higher-risk category and worse survival of MDS patients. By analyzing the MDS public database (GSE_58831) and q-PCR data of MDS patients in our center, we found that among 23 classical m6A related genes, m6A methyltransferase METTL14 was the key factor causing the m6A dysregulation in MDS: high expression of METTL14 was associated with higher disease risk stratification, more bone marrow blasts, poorer prognosis, and higher response rates to hypomethylating agents (HMAs). In order to explore the functions of METTL14 in MDS, we performed gain- and loss-of-function experiments in MDS cell line MDSL in vitro. It was observed that the expression of METTL14 promoted the cell proliferation and clone formation ability of MDS cells by inducing cell apoptosis and differentiation. More interestingly, we found that HMAs down-regulated the level of global m6A modification by inhibiting METTL14 expression, and the expression of METTL14 could increase the sensitivity of MDS cells to HMAs in MDS cells. To search for upstream regulatory genes of METTL14, we analyzed the ChIPBase online database and identified ELF1 might be the potential upstream transcription factor. The relationship between ELF1 and METTL14 was further confirmed by ELF1-overexpressed and ELF1-knowndowned MDSL cell lines. To explore the relationship between gene mutations and METTL14, we analyzed the correlations between next generation sequencing (NGS) data and METTL14 expression level in MDS patients in our center, and found that the METTL14 expression level was significantly increased in MDS patients carrying TP53 gene mutations (P<0.05). The increased METTL14 and ELF1 expression were all observed in MDS cell lines with introduction of TP53 missense mutations (M237I and Y220C) or TP53 knockdown. Mechanistically, m6A sequencing and RNA-seq sequencing revealed METTL14 promoted MDS development via PI3K/Akt signaling pathway, and the potential target genes involved in PI3K/Akt signaling pathway were ANGPT1, CDK6, FGFR2, GNG11, IGF1R et al. Conclusion: Herein, we reported RNA m6A methylation dysregulation did exist in MDS, and m6A methyltransferase METTL14 was an important regulatory factor. Higher METTL14 expression was associated with higher-risk stratification, poorer survival, and better response to HMAs in MDS patients. METTL14 was required for the cell growth and response to HMAs in MDS cells. TP53 mutations were significantly correlated with the up-regulation of METTL14 expression level, and TP53 mutations could induce METTL14 overexpression by regulating transcription factor ELF1. Mechanistically, METTL14 exerts its oncogenic role via PI3K/Akt signaling pathway. Our findings highlighted that METTL14 could potentially be a novel therapeutic target in MDS especially those with TP53 mutations. Figure 1View largeDownload PPTFigure 1View largeDownload PPT Close modal
-
METTL14-mediated epitranscriptome modification of MN1 mRNA promote tumorigenicity and all-trans-retinoic acid resistance in osteosarcoma
Hong-Bo Li,Gang Huang,Jian Tu,Dong-Ming Lv,Qing-Lin Jin,Jun-Kai Chen,Yu-Tong Zou,Dung-Fang Lee,Jing-Nan Shen,Xian-Biao Xie
DOI: https://doi.org/10.1016/j.ebiom.2022.104142
IF: 11.205
2022-08-01
EBioMedicine
Abstract:BACKGROUND: Osteosarcoma (OS) is the most common primary malignant bone tumor in adolescents. The molecular mechanism behind OS progression and metastasis remains poorly understood, which limits the effectiveness of current therapies. RNA N<sup>6</sup>-methyladenosine (m<sup>6</sup>A) modification plays a critical role in influencing RNA fate. However, the biological significance of m<sup>6</sup>A modification and its potential regulatory mechanisms in the development of OS remain unclear.METHODS: Liquid chromatography-tandem mass spectrometry (LC-MS/MS), dot blotting, and colorimetric ELISA were used to detect m<sup>6</sup>A levels. Western blotting, quantitative real-time PCR (RT-qPCR) and immunohistochemistry (IHC) were used to investigate METTL14 expression levels. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and transcriptomic RNA sequencing (RNA-seq) were used to screen the target genes of METTL14. RNA pull-down and RNA immunoprecipitation (RIP) assays were conducted to explore the specific binding of target genes and relevant m<sup>6</sup>A "readers". RNA stability and polysome analysis assays were used to detect the half-lives and translation efficiencies of the downstream genes of METTL14. IHC and clinical data were applied to explore the clinical correlations of METTL14 and its downstream target genes with the prognosis of OS.FINDINGS: We observed the abundance of m<sup>6</sup>A modifications in OS and revealed that METTL14 plays an oncogenic role in facilitating OS progression. MeRIP-seq and RNA-seq revealed that MN1 is a downstream gene of METTL14. MN1 contributes to tumor progression and all-trans-retinoic acid (ATRA) chemotherapy resistance in OS. Mechanistically, MN1 is methylated by METTL14, specifically in the coding sequence (CDS) regions, and this modification is recognized by the specific m<sup>6</sup>A reader insulin-like growth factor 2 mRNA binding protein 2 (IGF2BP2) to prevent MN1 mRNA degradation and promote it translation efficiency. IHC showed that MN1 expression was positively correlated with METTL14 and IGF2BP2 expression in OS tissues. The METTL14-IGF2BP2-MN1 panel demonstrated more promising prognostic value for OS patients than any of these molecules individually.INTERPRETATION: Our study revealed that METTL14 contributes to OS progression and ATRA resistance as an m<sup>6</sup>A RNA methylase by regulating the stability and translation efficiency of MN1 and thus provides both an underlying biomarker panel for prognosis prediction in OS patients.FUNDING: This work was supported by the National Natural Science Foundation of China (Grants 81972510 and 81772864).
medicine, research & experimental
-
Dysregulated m6A-Related Regulators Are Associated With Tumor Metastasis and Poor Prognosis in Osteosarcoma
Jianhao Li,Benchen Rao,Jie Yang,Liwen Liu,Maoxin Huang,Xin Liu,Guangying Cui,Chao Li,Qicai Han,Hao Yang,Xichun Cui,Ranran Sun
DOI: https://doi.org/10.3389/fonc.2020.00769
IF: 4.7
2020-06-02
Frontiers in Oncology
Abstract:Background: Osteosarcoma (OS) is the most common primary bone tumor. The disease has a poor prognosis due to the delay in the diagnosis and the development of metastasis. N6-Methyladenosine (m6A)-related regulators play an essential role in various tumors. In this study, a comprehensive analysis was conducted to elucidate the relationship between the expression profiles of m6A-related molecules and the clinical outcome of OS patients.Materials and Methods: Public genome datasets and a tissue microarray (TMA) cohort were used to analyze the mRNA and protein expression levels of m6A regulators. Next, immunofluorescence (IF) analysis was used to determine the subcellular localization of m6A-related molecules. Kaplan–Meier and Cox regression analyses were performed to confirm the prognostic value of m6A-related molecules in OS. A comprehensive bioinformatic analysis was conducted to identify the potential molecular mechanisms mediated by m6A modification in OS.Results: We found that m6A-related regulator expression was dysregulated in OS tissues, especially in metastatic tumor tissues. Low expression of METTL3, METTL14, and YTHDF2 and high expression of KIAA1429 and HNRNPA2B1 were significantly associated with poor prognosis in the TMA cohort. Simultaneously, the genome meta-cohort analysis revealed that low expression of FTO and METTL14 and high expression of METTL3, HNRNPA2B1, and YTHDF3 were associated with poor prognosis in OS. Cox regression analysis showed that HNRNPA2B1 might be an independent risk factor for OS. Bioinformatic analysis indicated that m6A regulators might be involved in OS progression through humoral immune response and cell cycle pathways.Conclusion: M6A-related regulators are frequently dysregulated and correlate with metastasis and prognosis in OS. M6A-related regulators may serve as novel therapeutic targets and prognostic biomarkers for OS.
oncology
-
Effects of RNA Methylation N6-methyladenosine Regulators on Malignant Progression and Prognosis of Melanoma.
Liu Jinfang,Zhou Zijian,Ma Ling,Li Chujun,Lin Yu,Yu Ting,Wei Ji-Fu,Zhu Lingjun,Yao Gang
DOI: https://doi.org/10.1186/s12935-021-02163-9
IF: 6.429
2021-01-01
Cancer Cell International
Abstract:BACKGROUND:Melanoma is an extremely aggressive type of skin cancer and experiencing a expeditiously rising mortality in a current year. Exploring new potential prognostic biomarkers and therapeutic targets of melanoma are urgently needed. The ambition of this research was to identify genetic markers and assess prognostic performance of N6-methyladenosine (m6A) regulators in melanoma.METHODS:Gene expression data and corresponding clinical informations of melanoma patients as well as sequence data of normal controls are collected from The Cancer Genome Atlas (TCGA) and the Genotype-Tissue Expression (GTEx) databases. Quantitative real-time PCR (qRT-PCR) analysis was carried out to detect the RNA expression of IGF2BP3 in A375 cell line, melanoma tissues, and normal tissues. Western blot, cell proliferation, and migration assays were performed to assess the ability of IGF2BP3 in A375 cell line.RESULTS:Differently expressed m6A regulators between tumor samples and normal samples were analyzed. A three-gene prognostic signature including IGF2BP3, RBM15B, and METTL16 was constructed, and the risk score of this signature was identified to be an independent prognostic indicator for melanoma. In addition, IGF2BP3 was verified to promote melanoma cell proliferation and migration in vitro and associate with lymph node metastasis in clinical samples. Moreover, risk score and the expression of IGF2BP3 were positively associated with the infiltrating immune cells and these hub genes made excellent potential drug targets in melanoma.CONCLUSION:We identified the genetic changes in m6A regulatory genes and constructed a three-gene risk signature with distinct prognostic value in melanoma. This research provided new insights into the epigenetic understanding of m6A regulators and novel therapeutic strategies in melanoma.
-
ZBTB7C m6A modification incurred by METTL3 aberration promotes osteosarcoma progression
Xueying An,Wenshu Wu,Lin Yang,Jian Dong,Bin Liu,Junxia Guo,Jianmei Chen,Baosheng Guo,Wangsen Cao,Qing Jiang
DOI: https://doi.org/10.1016/j.trsl.2023.04.005
IF: 10.171
2023-04-30
Translational Research
Abstract:Aberrant N6-methyladenosine (m6A) modification of mRNAs contributes significantly to the epigenetic tumorigenesis, however, its precise role and the key targets in osteosarcoma (OS) are not defined. Here we reported that selective METTL3 (methyltransferase like 3) elevation and the consequential increase of m6A modification causally affect OS progression. The fast-growing OS cells displayed preferential upregulation of METTL3 and increased m6A modification. Conversely, m6A inhibition by 3-deazaadenosine, siRNA-mediated METTL3 knockdown or a METTL3-selective inhibitor by STM2457 effectively inhibits OS cell growth and induced OS cell apoptosis. Further investigation revealed that an oncogenic protein ZBTB7C was likely a critical m6A target that mediated the oncogenic effects. ZBTB7C mRNA contains a typical m6A motif of high confidence and its mRNA and protein were enriched with increased m6A modification in OS samples/cells. In an OS xenograft model, STM2457 or siRNA-mediated METTL3 knockdown effectively lowed ZBTB7C abundance. More importantly, the anti-OS effects of STM2457 were significantly reduced when ZBTB7C was overexpressed by lentivirus. Together, our results demonstrate that the METTL3 aberration and the resultant ZBTB7C m6A modification form an important epigenetic regulatory loop that promotes OS progression, and targeting the METTL3/ZBTB7C axis may provide novel insights into the potential strategies for OS therapy.
medicine, general & internal,medical laboratory technology, research & experimental
-
Potential role of m6A RNA methylation regulators in osteosarcoma and its clinical prognostic value
Hua Liu,Guangzhen Qin,Yanan Ji,Xiaojian Wang,Hailin Bao,Xiaojun Guan,Aichun Wei,Zhigang Cai
DOI: https://doi.org/10.1186/s13018-021-02422-5
IF: 2.6
2021-05-05
Journal of Orthopaedic Surgery and Research
Abstract:Abstract Background Osteosarcoma is a disease with high mortality in children and adolescents, and metastasis is one of the important clinical features of osteosarcoma. N 6-Methyladenosine (m6A) is the most abundant methylation modification in mRNA, which is regulated by m6A regulators. It is reported that it is related to the occurrence and development of tumors. However, the mechanism of its action in osteosarcoma is rarely known. The purpose of this study was to identify the potential role of m6A regulatory factor in osteosarcoma and its clinical prognostic value. Methods Here, we used The Cancer Genome Atlas (TCGA) to comprehensively analyze the relationship between m6A regulatory factors and osteosarcoma (metastasis group and non-metastasis group). We analyzed their survival relationship and analyzed all the m6A regulatory factors in TCGA tumor data set by using the univariate Cox proportional hazard regression model. Finally, we selected two survival-related methylation regulators (FTO and IGF2BP2) as risk gene signature. Results According to the median risk, patients were divided into low-risk group and high-risk group. Multivariate Cox regression analysis showed that these two risk genes were considered to be the key factors independently predicting the prognosis of patients with osteosarcoma. In addition, we verified their characteristics with gene expression omnibus (GEO) DataSets and confirmed that they are related to tumor and immune-related signaling pathways through gene set enrichment analysis (GESA) and immune infiltration analysis. Conclusions In conclusion, m6A regulators might play an important role in the metastasis of osteosarcoma and have potential important value for the prognosis and treatment strategy of osteosarcoma patients.
orthopedics
-
N6-Methyladenosine-Related LncRNAs Are Potential Remodeling Indicators in the Tumor Microenvironment and Prognostic Markers in Osteosarcoma.
Zhongguang Wu,Xiaobo Zhang,Dongjie Chen,Zian Li,Xin Wu,Jianlong Wang,Youwen Deng
DOI: https://doi.org/10.3389/fimmu.2021.806189
IF: 7.3
2022-01-01
Frontiers in Immunology
Abstract:N6-Adenosine methylation, yielding N6-methyladenosine (m6A), is a reversible epigenetic modification found in messenger RNAs and long non-coding RNAs (lncRNAs), which affects the fate of modified RNA molecules and is essential for the development and differentiation of immune cells in the tumor microenvironment (TME). Osteosarcoma (OS) is the most common primary bone tumor in children and adolescents, and is characterized by high mortality. Currently, the possible role of m6A modifications in the prognosis of OS is unclear. In the present study, we investigated the correlation between m6A-related lncRNA expression and the clinical outcomes of OS patients via a comprehensive analysis. Clinical and workflow-type data were obtained from the Genotype-Tissue Expression Program and The Cancer Genome Atlas. We examined the relationship between m6A modifications and lncRNA expression, conducted Kyoto Encyclopedia of Genes analysis and also gene set enrichment analysis (GSEA), implemented survival analysis to investigate the association of clinical survival data with the expression of m6A-related lncRNAs, and utilized Lasso regression to model the prognosis of OS. Furthermore, we performed immune correlation analysis and TME differential analysis to investigate the infiltration levels of immune cells and their relationship with clinical prognosis. LncRNA expression and m6A levels were closely associated in co-expression analysis. The expression of m6A-related lncRNAs was quite low in tumor tissues; this appeared to be a predicting factor of OS in a prognostic model, independent of other clinical features. The NOD-like receptor signaling pathway was the most significantly enriched pathway in GSEA. In tumor tissues, SPAG4 was overexpressed while ZBTB32 and DEPTOR were downregulated. Tissues in cluster 2 were highly infiltrated by plasma cells. Cluster 2 presented higher ESTIMATE scores and stromal scores, showing a lower tumor cell purity in the TME. In conclusion, m6A-related lncRNA expression is strongly associated with the occurrence and development of OS, and can be used to as a prognostic factor of OS. Moreover, m6A-related lncRNAs and infiltrating immune cells in the TME could serve as new therapeutic targets and prognostic biomarkers for OS.
-
METTL1 Mediated m7G Modification Regulates Ferroptosis and Chemotherapy Resistance via Involvement of pri-miR-26a/FTH1 Axis in Osteosarcoma
Ye Yuan,Mingyu He,Yang Wang,Jiaying Pu,Zhihua Shen,Tong Wang,Guanghui Li,Zhongting Mei,Ao Wang,Zijing Ren,Ying Liu,Wenbo Wang,Xiaoyan Liu,Hong Lei,Xiaoqi He,Qian Liu,Jinhuan Hong,Weijie Du,Lei Yang
DOI: https://doi.org/10.21203/rs.3.rs-1003392/v1
2021-11-10
Abstract:Abstract Background N7-Methyladenosine (m7G), one of the most prevalent internal modifications in mammalian RNAs, makes a great contribution to many bioprocesses. However, its functions and underlying mechanisms in the occurrence and development of osteosarcoma remain elusive. Methods The expression of METTL1 and its correlation with clinicopathological features using tissue microarrays and the Cancer Genome Atlas (TCGA) dataset.Targets of METTL1 on osteosarcoma were identified by AlkAniline-seq, RNA immunoprecipitation (RIP), quantitative real-time PCR, western blot and luciferase assays. The effects of METTL1 on the biological characteristics of osteosarcoma cells were investigated on the basis of gain- and loss-of-function analyses. Ferroptosis were analyzed by lipid peroxidation assay, iron assay and ransmission electron microscopy (TEM). Subcutaneous models further uncovered the role of METTL1 in tumour growth. Results Here, we have identified that METTL1, an m7G methyltransferase, is low expressed in osteosarcoma tissues and plays an antitumor role in osteosarcoma. Mechanistically, METTL1 enhances cell ferroptosis by targeting pri-miR-26a and promoting its mature through m7G methylation, which could target FTH1 mRNA and eliminate FTH1 translation efficiency mediated by METTL1. FTH1 is the main component of ferritin which is crucial for iron homeostasis and the inhibition of lipid peroxidation. Reduction of FTH1 translation dramatically increases ferroptosis and enhances osteosarcoma chemosensitivity. Conclusions Collectively, our study demonstrates the METTL1/pri-miR-26a/FTH1 axis signaling in osteosarcoma and highlights the functional importance of METTL1 and m7G methylation in the progression and chemotherapy resistance of osteosarcoma, suggesting that reprogramming RNA m7G methylation as a potential and promising strategy for osteosarcoma treatment.
-
The m6A methyltransferase METTL3 promotes osteosarcoma progression by regulating the m6A level of LEF1
Wujun Miao,Jiajia Chen,Lianshun Jia,Jun Ma,Dianwen Song
DOI: https://doi.org/10.1016/j.bbrc.2019.06.128
2019-08-27
Abstract:Osteosarcoma(OS) is the most common and aggressive malignant bone sarcoma,which occurs in rapidly growing bones in children and adolescents. However, the underlying molecular mechanisms of OS development have not been fully illustrated. N6-Methyladenosine (m6A) is the most prevalent internal chemical modification of mRNAs, which is involved in many pathological processes in cancer development. However, its role and regulatory mechanism in OS remain unknown. In this study, we aimed to investigate the roles of m6A and its methyltransferase METTL3 in OS development. The results showed that m6A level for RNA methylation and the expression level of METTL3 were up-regulated in human OS tissues and OS cell lines. Functionally, lentivirus-mediated METTL3 silence in HOS and SAOS-2 cells inhibited the cell proliferation, migration and invasion ability. Further mechanism analysis suggested that METTL3 silence decreased the m6A methylation and total mRNA level of lymphoid enhancer-binding factor 1 (LEF1), followed by inhibited the activity of Wnt/β-catenin signaling pathway. Moreover, LEF1 over-expression abrogates the repressive effects of METTL3 silence on the proliferation, migration and invasion abilities of OS cells. Together, these results revealed that the m6A methyltransferase METTL3 promotes osteosarcoma cell progression by regulating the m6A level of LEF1 and activating Wnt/β-catenin signaling pathway.
-
Methylation recognition protein YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) regulates the proliferation, migration and invasion of osteosarcoma by regulating m6A level of CCR4-NOT transcription complex subunit 7 (CNOT7)
Kang Wei,Yi Gao,Bin Wang,Yu-Xing Qu
DOI: https://doi.org/10.1080/21655979.2022.2037381
2022-02-14
Bioengineered
Abstract:N6-methyladenosine (m6A) is one of the most significant modifications in human mRNAs. Emerging evidence indicates that m6A participates in the initiation and development of malignant tumors. Nevertheless, the biological roles and mechanism of m6A in osteosarcoma (OS) remain unclear. The purpose of this study was to investigate the role and mechanism of the methylation recognition protein-YTH N6-methyladenosine RNA binding protein 1 (YTHDF1) in OS. The YTHDF1 expression in OS was detected by qRT-PCR and Western blot assay. M6A quantification was utilized to measure the methylation level of OS. Cell counting kit-8 (CCK8), 5-Ethynyl-2'-deoxyuridine (EdU) assay and transwell experiments were conducted to confirm the biological effects of YTHDF1 on OS cells. The bioinformatics websites and in vitro assays were conducted to analyze the downstream targets of YTHDF1 was upregulated in OS tissues at mRNA and protein level. The results showed that the expression level of YTHDF1 might be closely associated with the poor prognosis for OS patients. Inhibition of YTHDF1 could suppress the proliferation, migration and invasion of the OS cells. Moreover, we found that CCR4-NOT transcription complex subunit 7 (CNOT7) might be the potential target of YTHDF1, which was upregulated in OS tissues. YTHDF1 could recognize the m6A sites of CONT7 and promote its expression in an m6A manner. Moreover, methyltransferase-like 3 (METTL3) could promote the m6A level of CONT7. YTHDF1 was upregulated in OS and could promote cell proliferation, migration and invasion. The METTL3-CONT7-YTHDF1 regulatory axis might be the potential target for the prognosis and therapy of OS.
biotechnology & applied microbiology
-
m6A methylation regulators and ncRNAs in osteosarcoma: Potential therapeutic strategies
Ce Shi,Lei Chen,Kui Huang,Guanghui Yang,Tingting Shi,Jinshuang Li,Hongbing Zheng
DOI: https://doi.org/10.1016/j.pbiomolbio.2024.10.003
IF: 4.799
2024-10-26
Progress in Biophysics and Molecular Biology
Abstract:Osteosarcoma (OS) represents the primary form of bone cancer observed in paediatric and adolescent populations. Nearly 10% to 15% of patients have metastases at diagnosis, and the 5-year survival rate was less than 20%. Although numerous investigators have offered significant efforts, the survival rates for patients with OS have remained almost unchanged over the past three decades. The most pervasive and abundant modification of internal transcripts in eukaryotic messenger RNAs (mRNAs) is N6-methyladenosine (m6A), and it is regulated by m6A methylation regulators. A number of recent studies have demonstrated that m6A modifications can regulate the biological activities of tumour cells and are intimately linked with cancer development, prognosis, drug resistance, and therapy. N6-methyladenosine modification of Non-coding RNA (ncRNA) has likewise shown a broad potential in gene regulation and tumor biology. Epigenetic changes induced by mRNAs and ncRNAs methylation are important for a better understanding of OS development and targeted drug development. Therefore, this paper summarises the biological functions of m6A-modified regulators in osteosarcoma and the role of mutual regulation between m6A and ncRNAs in osteosarcoma. Furthermore, the potential clinical applications of m6A modifications in OS are presented for consideration. It provides new directions for the future research and clinical treatment strategies of osteosarcoma.
biochemistry & molecular biology,biophysics
-
m6A methylation modification: Potential pathways to suppress osteosarcoma metastasis
Tianrui Hu,Guowei Wang,Dong Wang,Youwen Deng,Weiguo Wang
DOI: https://doi.org/10.1016/j.intimp.2024.113759
IF: 5.714
2024-12-12
International Immunopharmacology
Abstract:Osteosarcoma is a highly aggressive malignant bone tumor prone to metastasis, and its metastatic process is one of the main reasons for treatment failure and poor prognosis. Recent studies have demonstrated that modification of m6A methylation plays an important role in osteosarcoma metastasis, influencing both invasion and metastasis through various signaling pathways. Therefore, clarification of the specific effects of m6A methylation modification in osteosarcoma may reveal ways to improve the prognosis of osteosarcoma patients. The roles of various components involved in the m6A methylation modification process in osteosarcoma have been investigated, with studies focusing more on their effects than on their mechanisms. In this review, we focus on the interactions between the "writers," "erasers," and "readers" of m6A methylation and tumor metastasis-related factors to enhance the understanding of osteosarcoma and m6A methylation modification, with the aim of identifying clinical diagnostic biomarkers and potential therapeutic targets for osteosarcoma metastasis.
pharmacology & pharmacy,immunology
-
m6A regulators are associated with osteosarcoma metastasis and have prognostic significance: A study based on public databases
Wenpeng Zhang,Lina Wang,Ping Zhang,Quanbin Zhang
DOI: https://doi.org/10.1097/MD.0000000000025952
2021-05-21
Abstract:Background: Osteosarcoma represents the most common malignant bone tumor with high metastatic potential and inferior prognosis. RNA methylation (N6-methyladenosine [m6A]) is a prevalent RNA modification that epigenetically influences numerous biological processes including tumorigenesis. This study aims to determine that m6A regulators are significant biomarkers for osteosarcoma, and establish a prognostic model to predict the survival of patients. Methods: In this study, we comprehensively analyzed the underlying associations between m6A regulators' mRNA expressions and metastasis as well as prognosis of osteosarcoma patients in the Cancer Genome Atlas. Multivariate Cox-regression analysis was used to screen regulators that were significantly associated with overall survival of osteosarcoma patients. Least absolute shrinkage and selection operator (LASSO) Cox-regression analysis was used for constructing m6A regulator-based osteosarcoma prognostic signature. Results: Some of the regulators exhibited aberrant mRNA levels between osteosarcoma samples with and without metastasis. Multivariate Cox-regression analysis identified several regulators with potential prognostic significance. A risk score formula consisted of methyltransferase-like 3, YTH domains of Homo sapiens, and fat mass and obesity-associated protein was obtained through which patients could be prognostically stratified independently of potential confounding factors. The signature was also significantly associated with the metastatic potential of osteosarcoma. All the analyses could be well reproduced in another independent osteosarcoma cohort from the Gene Expression Omnibus. Conclusions: In conclusion, this study first revealed potential roles of m6A regulators in osteosarcoma metastasis and prognosis, which should be helpful for its clinical decision-making.
-
m6A-dependent upregulation of TRAF6 by METTL3 is associated with metastatic osteosarcoma
Jing Wang,Wentao Wang,Xing Huang,Jiashi Cao,Shuming Hou,Xiangzhi Ni,Cheng Peng,Tielong Liu
DOI: https://doi.org/10.1016/j.jbo.2022.100411
IF: 4.491
2022-02-01
Journal of Bone Oncology
Abstract:OBJECTIVES: RNA N6-methyladenosine (m6A) is associated with tumorigenesis. The importance of methyltransferase-like 3 (METTL3) has been reported in cancer progression and metastasis. However, its role and molecular mechanism in osteosarcoma (OS), the most common primary bone tumor, is poorly studied. In this study, we aimed to investigate the functional role and underlying mechanism of METTL3 in the metastasis of OS.METHODS: The expression differences of METTL3 between metastatic and non-metastatic OS tissues and patients with different Enneking stages were detected using RT-qPCR. METTL3 was artificially downregulated in the cells, followed by wound healing assay, Matrigel assay, immunofluorescence, <i>in vivo</i> tumorigenic assay, HE staining, and western blot. Transcriptome sequencing and m6A-seq was conducted to identify the downstream genes of METTL3, and RIP and dual-luciferase assays were performed for validation. The expression of TRAF6 in OS tissues was detected using RT-qPCR. Finally, the rescue experiments were conducted.RESULTS: METTL3 was overexpressed in metastatic OS tissues, and downregulation of METTL3 decreased cell migration, invasion, epithelial-mesenchymal transition, and tumorigenic and metastatic activities. The m6A site was highly enriched in cells poorly expressing METTL3, and the m6A peak was mainly enriched in the exon region. METTL3 was positively correlated with TRAF6 in metastatic OS, and depletion of METTL3 resulted in the loss of TRAF6 expression in OS cells. Upregulation of TRAF6 contributed to metastases <i>in vitro</i> and <i>in vivo</i>.CONCLUSION: METTL3 is highly expressed in OS and enhances TRAF6 expression through m6A modification, thereby promoting the metastases of OS cells.
oncology
-
METTL16 promotes osteosarcoma progression by downregulating VPS33B in an m6A‐dependent manner
Jun Cheng,Zhihao Xu,Wei Tan,Jinpeng He,Boyu Pan,Yan Zhang,Youwen Deng
DOI: https://doi.org/10.1002/jcp.31068
2023-06-26
Journal of Cellular Physiology
Abstract:This research article aims to investigate the role of methyltransferase‐like 16 (METTL16) on osteosarcoma. It reveals that METTL16 regulates the progression of osteosarcoma by impairing vacuolar protein sorting protein 33b (VPS33B) messenger RNA via YTHDF1. Moreover, METTL16/VPS33B axi promotes osteosarcoma progression via PI3K/AKT pathway. N6‐methyladenosine (m6A) is one of the main epitranscriptomic modifications that accelerates the progression of malignant tumors by modifying RNA. Methyltransferase‐like 16 (METTL16) is a newly identified methyltransferase that has been found to play an important oncogenic role in a few malignancies; however, its function in osteosarcoma (OS) remains unclear. In this study, METTL16 was found to be upregulated in OS tissues, and associated with poor prognosis in OS patients. Functionally, METTL16 substantially promoted OS cell proliferation, migration, and invasion in vitro and OS growth in vivo. Mechanistically, vacuolar protein sorting protein 33b (VPS33B) was identified as the downstream target of METTL16, which induced m6A modification of VPS33B and impaired the stability of the VPS33B transcript, thereby degrading VPS33B. In addition, VPS33B was found to be downregulated in OS tissues, VPS33B knockdown markedly attenuated shMETTL16‐mediated inhibition on OS progression. Finally, METTL16/VPS33B might facilitate OS progression through PI3K/AKT pathway. In summary, this study revealed an important role for the METTL16‐mediated m6A modification in OS progression, implying it as a promising target for OS treatment.
cell biology,physiology
-
The Emerging Molecular Mechanism of M6a Modulators in Tumorigenesis and Cancer Progression
Shuiping Liu,Qiujie Li,Ke Chen,Qin Zhang,Guohua Li,Lvjia Zhuo,Bingtao Zhai,Xinbing Sui,Xiaotong Hu,Tian Xie
DOI: https://doi.org/10.1016/j.biopha.2020.110098
IF: 7.419
2020-01-01
Biomedicine & Pharmacotherapy
Abstract:N6-methyladenosine (m6A) is the most abundant RNA modification; m6A modifications are installed by methyltransferases, removed by demethylases and recognized by reader proteins. M6A plays crucial roles in a variety of biological processes by regulating target RNA translation, splicing, nuclear export, and decay. Since the establishment of methylated RNA immunoprecipitation-sequencing methodology, over three hundred articles about m6A modulators, including "writers", "erasers" and "readers", have been reported in the last four years. In addition, an increasing number of molecular mechanisms underlying m6A RNA methylation in human cancers have been comprehensively clarified. The recently emerged molecular mechanisms of m6A modulators in cancer cell proliferation, cell cycle progression, migration and invasion, apoptosis, and autophagy remain to be summarized. Hence, this review specifically summarizes these recent advances in the understanding of m6A molecular mechanisms in tumorigenesis and cancer progression. In addition, we discuss the prospect of using an m6A methylation modulator as a new diagnostic biomarker and therapeutic target for human cancers.
-
M6A demethylase FTO-mediated downregulation of DACT1 mRNA stability promotes Wnt signaling to facilitate osteosarcoma progression
Dongming Lv,Shirong Ding,Li Zhong,Jian Tu,Hongbo Li,Hao Yao,Yutong Zou,Ziliang Zeng,Yan Liao,Xuesi Wan,Lili Wen,Xianbiao Xie
DOI: https://doi.org/10.1038/s41388-022-02214-z
IF: 8.756
2022-02-04
Oncogene
Abstract:Despite advances in clinical diagnosis and treatment, the prognosis of patients with osteosarcoma (OS) remains poor, and the treatment efficacy has plateaued. Therefore, it is important to identify new therapeutic targets for OS. N6-methyladenosine (m6A) modification has been reported to participate in tumor malignancy. In this study, functional screening showed that the m6A demethylase FTO could be a candidate therapeutic target for OS. Upregulated FTO in OS could predict a poorer prognosis. FTO promoted the growth and metastasis of OS in vitro and in vivo. Methylated RNA immunoprecipitation sequencing (MeRIP-seq) and RNA sequencing (RNA-seq) were performed to identify DACT1 as a potential target of FTO. In vitro assays demonstrated that FTO could reduce the mRNA stability of DACT1 via m6A demethylation, which decreased DACT1 expression and further activated the Wnt signaling pathway. The oncogenic effect of FTO on OS was dependent on DACT1. In addition, the m6A reader IGF2BP1 was validated to participate in the regulation of DACT1. Entacapone, a conventional drug for Parkinson’s disease, was confirmed to suppress OS via m6A-mediated regulation through the FTO/DACT1 axis. Our findings demonstrate that FTO may be a novel therapeutic target and that entacapone has preclinical value to be repurposed for OS.
oncology,genetics & heredity,biochemistry & molecular biology,cell biology
-
M 7 G modification of FTH1 and pri-miR-26a regulates ferroptosis and chemotherapy resistance in osteosarcoma
Mingyu He,Yang Wang,Jiajie Xie,Jiaying Pu,Zhihua Shen,Ao Wang,Tao Li,Tong Wang,Guanghui Li,Ying Liu,Zhongting Mei,Zijing Ren,Wenbo Wang,Xiaoyan Liu,Jinhuan Hong,Qian Liu,Hong Lei,Xiaoqi He,Weijie Du,Ye Yuan,Lei Yang
DOI: https://doi.org/10.1038/s41388-023-02882-5
IF: 8.756
2023-12-01
Oncogene
Abstract:Doxorubicin and platinum are widely used in the frontline treatment of osteosarcoma, but resistance to chemotherapy limits its curative effect. Here, we have identified that METTL1 mediated N 7 -Methyladenosine (m 7 G) low expressed in osteosarcoma tissues, plays a critical oncogenic role, and enhances osteosarcoma chemosensitivity in osteosarcoma. Mechanistically, AlkAniline-Seq data revealed that Ferritin heavy chain (FTH1), the main component of ferritin, which is crucial for iron homeostasis and the inhibition of lipid peroxidation, is one of the top 10 genes with the most significant change in m 7 G methylation sites mediated by METTL1 in human osteosarcoma cells. Interestingly, METTL1 significantly increased the expression of FTH1 at the mRNA level but was remarkably suppressed at the protein level. We then identified primary (pri)-miR-26a and pri-miR-98 in the Top 20 m 7 G-methylated pri-miRNAs with highly conserved species. Further results confirmed that METTL1 enhances cell ferroptosis by targeting FTH1 and primary (pri)-miR-26a, promoting their maturity by enhancing RNA stability dependent on m 7 G methylation. The increase of mature miR-26a-5p that resulted from METTL1 overexpression could further target FTH1 mRNA and eliminate FTH1 translation efficiency. Moreover, the reduction of FTH1 translation dramatically increases cell ferroptosis and promotes the sensitivity of osteosarcoma cells to chemotherapy drugs. Collectively, our study demonstrates the METTL1/pri-miR-26a/FTH1 axis signaling in osteosarcoma and highlights the functional importance of METTL1 and m 7 G methylation in the progression and chemotherapy resistance of osteosarcoma, suggesting that reprogramming RNA m 7 G methylation as a potential and promising strategy for osteosarcoma treatment.
oncology,genetics & heredity,biochemistry & molecular biology,cell biology
-
METTL16 promotes osteosarcoma progression by downregulating VPS33B in an m6 A-dependent manner
Jun Cheng,Zhihao Xu,Wei Tan,Jinpeng He,Boyu Pan,Yan Zhang,Youwen Deng
DOI: https://doi.org/10.1002/jcp.31068
Abstract:N6-methyladenosine (m6 A) is one of the main epitranscriptomic modifications that accelerates the progression of malignant tumors by modifying RNA. Methyltransferase-like 16 (METTL16) is a newly identified methyltransferase that has been found to play an important oncogenic role in a few malignancies; however, its function in osteosarcoma (OS) remains unclear. In this study, METTL16 was found to be upregulated in OS tissues, and associated with poor prognosis in OS patients. Functionally, METTL16 substantially promoted OS cell proliferation, migration, and invasion in vitro and OS growth in vivo. Mechanistically, vacuolar protein sorting protein 33b (VPS33B) was identified as the downstream target of METTL16, which induced m6 A modification of VPS33B and impaired the stability of the VPS33B transcript, thereby degrading VPS33B. In addition, VPS33B was found to be downregulated in OS tissues, VPS33B knockdown markedly attenuated shMETTL16-mediated inhibition on OS progression. Finally, METTL16/VPS33B might facilitate OS progression through PI3K/AKT pathway. In summary, this study revealed an important role for the METTL16-mediated m6 A modification in OS progression, implying it as a promising target for OS treatment.
-
The Emerging Roles of RNA m6A Methylation and Demethylation as Critical Regulators of Tumorigenesis, Drug Sensitivity, and Resistance
Qing Lan,Pei Y. Liu,Jessica L. Bell,Jenny Y. Wang,Stefan Hüttelmaier,Xu Dong Zhang,Lirong Zhang,Tao Liu
DOI: https://doi.org/10.1158/0008-5472.can-20-4107
IF: 11.2
2021-03-05
Cancer Research
Abstract:Abstract RNA N6-methyladenosine (m6A) modification occurs in approximately 25% of mRNAs at the transcriptome-wide level. RNA m6A is regulated by the RNA m6A methyltransferases methyltransferase-like 3 (METTL3), METTL14, and METTL16 (writers), demethylases FTO and ALKBH5 (erasers), and binding proteins YTHDC1–2, YTHDF1–3, IGF2BP1–3, and SND1 (readers). These RNA m6A modification proteins are frequently upregulated or downregulated in human cancer tissues and are often associated with poor patient prognosis. By modulating pre-mRNA splicing, mRNA nuclear export, decay, stability, and translation of oncogenic and tumor suppressive transcripts, RNA m6A modification proteins regulate cancer cell proliferation, survival, migration, invasion, tumor initiation, progression, metastasis, and sensitivity to anticancer therapies. Importantly, small-molecule activators of METTL3, as well as inhibitors of METTL3, FTO, ALKBH5, and IGF2BP1 have recently been identified and have shown considerable anticancer effects when administered alone or in combination with other anticancer agents, both in vitro and in mouse models of human cancers. Future compound screening and design of more potent and selective RNA m6A modification protein inhibitors and activators are expected to provide novel anticancer agents, appropriate for clinical trials in patients with cancer tissues harboring aberrant RNA m6A modification protein expression or RNA m6A modification protein–induced resistance to cancer therapy.
oncology